1
|
Pun R, North BJ. Role of spindle assembly checkpoint proteins in gametogenesis and embryogenesis. Front Cell Dev Biol 2025; 12:1491394. [PMID: 39911185 PMCID: PMC11794522 DOI: 10.3389/fcell.2024.1491394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 12/17/2024] [Indexed: 02/07/2025] Open
Abstract
The spindle assembly checkpoint (SAC) is a surveillance mechanism that prevents uneven segregation of sister chromatids between daughter cells during anaphase. This essential regulatory checkpoint prevents aneuploidy which can lead to various congenital defects observed in newborns. Many studies have been carried out to elucidate the role of proteins involved in the SAC as well as the function of the checkpoint during gametogenesis and embryogenesis. In this review, we discuss the role of SAC proteins in regulating both meiotic and mitotic cell division along with several factors that influence the SAC strength in various species. Finally, we outline the role of SAC proteins and the consequences of their absence or insufficiency on proper gametogenesis and embryogenesis in vivo.
Collapse
Affiliation(s)
| | - Brian J. North
- Biomedical Sciences Department, School of Medicine, Creighton University, Omaha, NE, United States
| |
Collapse
|
2
|
Ballmer D, Lou HJ, Ishii M, Turk BE, Akiyoshi B. Aurora B controls anaphase onset and error-free chromosome segregation in trypanosomes. J Cell Biol 2024; 223:e202401169. [PMID: 39196069 PMCID: PMC11354203 DOI: 10.1083/jcb.202401169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/12/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Kinetochores form the interface between chromosomes and spindle microtubules and are thus under tight control by a complex regulatory circuitry. The Aurora B kinase plays a central role within this circuitry by destabilizing improper kinetochore-microtubule attachments and relaying the attachment status to the spindle assembly checkpoint. Intriguingly, Aurora B is conserved even in kinetoplastids, a group of early-branching eukaryotes which possess a unique set of kinetochore proteins. It remains unclear how their kinetochores are regulated to ensure faithful chromosome segregation. Here, we show in Trypanosoma brucei that Aurora B activity controls the metaphase-to-anaphase transition through phosphorylation of the divergent Bub1-like protein KKT14. Depletion of KKT14 overrides the metaphase arrest resulting from Aurora B inhibition, while expression of non-phosphorylatable KKT14 delays anaphase onset. Finally, we demonstrate that re-targeting Aurora B to the outer kinetochore suffices to promote mitotic exit but causes extensive chromosome missegregation in anaphase. Our results indicate that Aurora B and KKT14 are involved in an unconventional circuitry controlling cell cycle progression in trypanosomes.
Collapse
Affiliation(s)
- Daniel Ballmer
- Department of Biochemistry, University of Oxford, Oxford, UK
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Hua Jane Lou
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Midori Ishii
- Department of Biochemistry, University of Oxford, Oxford, UK
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Benjamin E. Turk
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Bungo Akiyoshi
- Department of Biochemistry, University of Oxford, Oxford, UK
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
3
|
Koo ASH, Jia W, Kim SH, Scalf M, Boos CE, Chen Y, Wang D, Voter AF, Bajaj A, Smith LM, Keck JL, Bakkenist CJ, Guo L, Tibbetts RS. Alternative splicing modulates chromatin interactome and phase separation of the RIF1 C-terminal domain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.619708. [PMID: 39553946 PMCID: PMC11565852 DOI: 10.1101/2024.10.29.619708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
RIF1 (RAP1 interacting factor) fulfills diverse roles in DNA double-strand break repair, DNA replication, and nuclear organization. RIF1 is expressed as two splice variants, RIF1-Long (RIF1-L) and RIF1-Short (RIF1-S), from the alternative splicing (AS) of Exon 32 (Ex32) which encodes a 26 aa Ser/Lys-rich cassette peptide in the RIF1 C-terminal domain (CTD). Here we demonstrate that Ex32 inclusion was repressed by DNA damage and oncogenesis but peaked at G2/M phase of the cell cycle. Ex32 splice-in was catalyzed by positive regulators including SRSF1, which bound to Ex32 directly, and negative regulators such as PTBP1 and SRSF3. Isoform proteomics revealed enhanced association of RIF1-L with MDC1, whose recruitment to IR-induced foci was strengthened by RIF1-L. RIF1-L and RIF1-S also exhibited unique phase separation and chromatin-binding characteristics that were regulated by CDK1-dependent CTD phosphorylation. These combined findings suggest that regulated AS affects multiple aspects of RIF1 function in genome protection and organization.
Collapse
Affiliation(s)
- Adenine Si-Hui Koo
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, Madison, WI 53705, USA
| | - Weiyan Jia
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, Madison, WI 53705, USA
| | - Sang Hwa Kim
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, Madison, WI 53705, USA
| | - Mark Scalf
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Ave, Madison, WI 53706, USA
| | - Claire E. Boos
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Ave, Madison, WI 53706, USA
| | - Yuhong Chen
- Versiti Blood Research Institute, Milwaukee, WI, 53226, USA
| | - Demin Wang
- Versiti Blood Research Institute, Milwaukee, WI, 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Andrew F. Voter
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, 420 Henry Mall, Madison, WI 53706, USA
| | - Aditya Bajaj
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Lloyd M. Smith
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Ave, Madison, WI 53706, USA
| | - James L. Keck
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, 420 Henry Mall, Madison, WI 53706, USA
| | | | - Lin Guo
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Randal S. Tibbetts
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, Madison, WI 53705, USA
| |
Collapse
|
4
|
Chen C, Li P, Fan G, Yang E, Jing S, Shi Y, Gong Y, Zhang L, Wang Z. Role of TRIP13 in human cancer development. Mol Biol Rep 2024; 51:1088. [PMID: 39436503 DOI: 10.1007/s11033-024-10012-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024]
Abstract
As an AAA + ATPase, thyroid hormone receptor interacting protein 13 (TRIP13) primarily functions in DNA double-strand break repair, chromosome recombination, and cell cycle checkpoint regulation; aberrant expression of TRIP13 can result in chromosomal instability (CIN). According to recent research, TRIP13 is aberrantly expressed in a variety of cancers, and a patient's poor prognosis and tumor stage are strongly correlated with high expression of TRIP13. Tumor cell and subcutaneous xenograft growth can be markedly inhibited by TRIP13 knockdown or TRIP13 inhibitor administration. In the initiation and advancement of human malignancies, TRIP13 seems to function as an oncogene. Based on available data, TRIP13 may function as a biological target and biomarker for cancer. The creation of inhibitors that specifically target TRIP13 may present novel approaches to treating cancer.
Collapse
Affiliation(s)
- Chaohu Chen
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Pan Li
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Guangrui Fan
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Enguang Yang
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Suoshi Jing
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Yibo Shi
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Yuwen Gong
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Luyang Zhang
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Zhiping Wang
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China.
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China.
| |
Collapse
|
5
|
Sun L, Chen X, Song C, Shi W, Liu L, Bai S, Wang X, Chen J, Jiang C, Wang SM, Luo ZQ, Wang R, Wang Y, Jin QW. Negative regulation of APC/C activation by MAPK-mediated attenuation of Cdc20 Slp1 under stress. eLife 2024; 13:RP97896. [PMID: 39412391 PMCID: PMC11483130 DOI: 10.7554/elife.97896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Mitotic anaphase onset is a key cellular process tightly regulated by multiple kinases. The involvement of mitogen-activated protein kinases (MAPKs) in this process has been established in Xenopus egg extracts. However, the detailed regulatory cascade remains elusive, and it is also unknown whether the MAPK-dependent mitotic regulation is evolutionarily conserved in the single-cell eukaryotic organisms such as fission yeast (Schizosaccharomyces pombe). Here, we show that two MAPKs in S. pombe indeed act in concert to restrain anaphase-promoting complex/cyclosome (APC/C) activity upon activation of the spindle assembly checkpoint (SAC). One MAPK, Pmk1, binds to and phosphorylates Slp1Cdc20, the co-activator of APC/C. Phosphorylation of Slp1Cdc20 by Pmk1, but not by Cdk1, promotes its subsequent ubiquitylation and degradation. Intriguingly, Pmk1-mediated phosphorylation event is also required to sustain SAC under environmental stress. Thus, our study establishes a new underlying molecular mechanism of negative regulation of APC/C by MAPK upon stress stimuli, and provides a previously unappreciated framework for regulation of anaphase entry in eukaryotic cells.
Collapse
Affiliation(s)
- Li Sun
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen UniversityXiamenChina
| | - Xuejin Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen UniversityXiamenChina
| | - Chunlin Song
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen UniversityXiamenChina
| | - Wenjing Shi
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen UniversityXiamenChina
| | - Libo Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen UniversityXiamenChina
| | - Shuang Bai
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen UniversityXiamenChina
| | - Xi Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen UniversityXiamenChina
| | - Jiali Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen UniversityXiamenChina
| | - Chengyu Jiang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen UniversityXiamenChina
| | - Shuang-min Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen UniversityXiamenChina
| | - Zhou-qing Luo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen UniversityXiamenChina
| | - Ruiwen Wang
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou UniversityFuzhouChina
| | - Yamei Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen UniversityXiamenChina
| | - Quan-wen Jin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen UniversityXiamenChina
| |
Collapse
|
6
|
Vazquez-Fernandez E, Yang J, Zhang Z, Andreeva AE, Emsley P, Barford D. A comparative study of the cryo-EM structures of Saccharomyces cerevisiae and human anaphase-promoting complex/cyclosome (APC/C). eLife 2024; 13:RP100821. [PMID: 39401078 PMCID: PMC11473103 DOI: 10.7554/elife.100821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024] Open
Abstract
The anaphase-promoting complex/cyclosome (APC/C) is a large multi-subunit E3 ubiquitin ligase that controls progression through the cell cycle by orchestrating the timely proteolysis of mitotic cyclins and other cell cycle regulatory proteins. Although structures of multiple human APC/C complexes have been extensively studied over the past decade, the Saccharomyces cerevisiae APC/C has been less extensively investigated. Here, we describe medium resolution structures of three S. cerevisiae APC/C complexes: unphosphorylated apo-APC/C and the ternary APC/CCDH1-substrate complex, and phosphorylated apo-APC/C. Whereas the overall architectures of human and S. cerevisiae APC/C are conserved, as well as the mechanism of CDH1 inhibition by CDK-phosphorylation, specific variations exist, including striking differences in the mechanism of coactivator-mediated stimulation of E2 binding, and the activation of APC/CCDC20 by phosphorylation. In contrast to human APC/C in which coactivator induces a conformational change of the catalytic module APC2:APC11 to allow E2 binding, in S. cerevisiae apo-APC/C the catalytic module is already positioned to bind E2. Furthermore, we find no evidence of a phospho-regulatable auto-inhibitory segment of APC1, that in the unphosphorylated human APC/C, sterically blocks the CDC20C-box binding site of APC8. Thus, although the functions of APC/C are conserved from S. cerevisiae to humans, molecular details relating to their regulatory mechanisms differ.
Collapse
Affiliation(s)
| | - Jing Yang
- MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | - Ziguo Zhang
- MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | | | - Paul Emsley
- MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | - David Barford
- MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
| |
Collapse
|
7
|
Ejaz U, Dou Z, Yao PY, Wang Z, Liu X, Yao X. Chromothripsis: an emerging crossroad from aberrant mitosis to therapeutic opportunities. J Mol Cell Biol 2024; 16:mjae016. [PMID: 38710586 PMCID: PMC11487160 DOI: 10.1093/jmcb/mjae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/23/2024] [Accepted: 05/04/2024] [Indexed: 05/08/2024] Open
Abstract
Chromothripsis, a type of complex chromosomal rearrangement originally known as chromoanagenesis, has been a subject of extensive investigation due to its potential role in various diseases, particularly cancer. Chromothripsis involves the rapid acquisition of tens to hundreds of structural rearrangements within a short period, leading to complex alterations in one or a few chromosomes. This phenomenon is triggered by chromosome mis-segregation during mitosis. Errors in accurate chromosome segregation lead to formation of aberrant structural entities such as micronuclei or chromatin bridges. The association between chromothripsis and cancer has attracted significant interest, with potential implications for tumorigenesis and disease prognosis. This review aims to explore the intricate mechanisms and consequences of chromothripsis, with a specific focus on its association with mitotic perturbations. Herein, we discuss a comprehensive analysis of crucial molecular entities and pathways, exploring the intricate roles of the CIP2A-TOPBP1 complex, micronuclei formation, chromatin bridge processing, DNA damage repair, and mitotic checkpoints. Moreover, the review will highlight recent advancements in identifying potential therapeutic targets and the underlying molecular mechanisms associated with chromothripsis, paving the way for future therapeutic interventions in various diseases.
Collapse
Affiliation(s)
- Umer Ejaz
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Chemical Biology, Hefei National Science Center for Inter-disciplinary Sciences, Hefei 230027, China
| | - Zhen Dou
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Chemical Biology, Hefei National Science Center for Inter-disciplinary Sciences, Hefei 230027, China
| | - Phil Y Yao
- University of California San Diego School of Medicine, San Diego, CA 92103, USA
| | - Zhikai Wang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Chemical Biology, Hefei National Science Center for Inter-disciplinary Sciences, Hefei 230027, China
| | - Xing Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Chemical Biology, Hefei National Science Center for Inter-disciplinary Sciences, Hefei 230027, China
| | - Xuebiao Yao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
| |
Collapse
|
8
|
Lebeau G, Hoareau M, Rivière S, El Safadi D, Da Silva CR, Krejbich-Trotot P, Viranaicken W. Cell cycle and mitosis progression during ZIKA virus infection: The viral non-structural protein NS5 as a master regulator of the APC/cyclosome? Biochimie 2024; 221:75-80. [PMID: 38307244 DOI: 10.1016/j.biochi.2024.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 01/15/2024] [Accepted: 01/30/2024] [Indexed: 02/04/2024]
Abstract
Alterations in cell cycle regulation contribute to Zika virus (ZIKV)-associated pathogenesis and may have implications for the development of therapeutic avenues. As a matter of fact, ZIKV alters cell cycle progression at multiple stages, including G1, S, G2, and M phases. During a cell cycle, the progression of mitosis is particularly controlled to avoid any abnormalities in cell division. In this regard, the critical metaphase-anaphase transition is triggered by the activation of anaphase-promoting complex/cyclosome (APC/C) by its E3 ubiquitin ligase subunit Cdc20. Cdc20 recognizes substrates by interacting with a destruction box motif (D-box). Recently, the ZIKV nonstructural protein 5 (NS5), one of the most highly conserved flavivirus proteins, has been shown to localize to the centrosome in each pole and to spindle fibers during mitosis. Inducible expression of NS5 reveals an interaction of this viral factor with centrosomal proteins leading to an increase in the time required to complete mitosis. By analyzing the NS5 sequence, we discovered the presence of a D-box. Taken together, these data support the idea that, in addition to its role in viral replication, NS5 plays a critical role in the control of the cell cycle of infected cells and, more specifically, in the regulation of the mitotic spindle. Here we propose that the NS5 protein may interfere with the metaphase-anaphase progression, and thus cause the observed delay in mitosis via the regulation of APC/C.
Collapse
Affiliation(s)
- Grégorie Lebeau
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791, Sainte Clotilde, La Réunion, France
| | - Mathilde Hoareau
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791, Sainte Clotilde, La Réunion, France; Université de La Réunion, INSERM, UMR 1188 Diabète athérothombose Réunion Océan Indien (DéTROI), 97410, Saint-Pierre, France
| | - Sébastien Rivière
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791, Sainte Clotilde, La Réunion, France; Université de La Réunion, INSERM, UMR 1188 Diabète athérothombose Réunion Océan Indien (DéTROI), 97410, Saint-Pierre, France
| | - Daed El Safadi
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791, Sainte Clotilde, La Réunion, France
| | - Christine Robert Da Silva
- Université de La Réunion, INSERM, UMR 1188 Diabète athérothombose Réunion Océan Indien (DéTROI), 97410, Saint-Pierre, France
| | - Pascale Krejbich-Trotot
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791, Sainte Clotilde, La Réunion, France.
| | - Wildriss Viranaicken
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791, Sainte Clotilde, La Réunion, France; Université de La Réunion, INSERM, UMR 1188 Diabète athérothombose Réunion Océan Indien (DéTROI), 97410, Saint-Pierre, France.
| |
Collapse
|
9
|
Ma T, Zhou S, Xie X, Chen J, Wang J, Zhang G. A case report of a family with developmental arrest of human prokaryotic stage zygote. Front Cell Dev Biol 2024; 12:1280797. [PMID: 38606321 PMCID: PMC11006971 DOI: 10.3389/fcell.2024.1280797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 02/19/2024] [Indexed: 04/13/2024] Open
Abstract
To study the genetic variation leading to the arrest phenotype of pronuclear (PN) zygotes. We recruited a family characterized by recurrent PN arrest during in vitro fertilization (IVF) and intracytoplasmic sperm injection cycles (ICSI) and performed whole-exome sequencing for 2 individuals. The transcriptome profiles of PN-arrest zygotes were assessed by single-cell RNA sequencing analysis. The variants were then validated by PCR amplification and Sanger sequencing in the affected individuals and other family members. A family characterized by recurrent PN arrest during IVF and ICSI cycles were enrolled after giving written informed consent. Peripheral blood samples were taken for DNA extraction. Three PN-arrest zygotes from patient III-3 were used for single-cell RNA-seq as described. This phenotype was reproduced after multiple cycles of egg retrieval and after trying different fertilization methods and multiple ovulation regimens. The mutant genes of whole exon sequencing were screened and verified. The missense variant c. C1630T (p.R544W) in RGS12 was responsible for a phenotype characterized by paternal transmission. RGS12 controls Ca2+ oscillation, which is required for oocyte activation after fertilization. Single-cell transcriptome profiling of PN-arrest zygotes revealed defective established translation, RNA processing and cell cycle, which explained the failure of complete oocyte activation. Furthermore, we identified proximal genes involved in Ca2+ oscillation-cytostatic factor-anaphase-promoting complex (Ca2+ oscillation-CSF-APC) signaling, including upregulated CaMKII, ORAI1, CDC20, and CDH1 and downregulated EMI1 and BUB3. The findings indicate abnormal spontaneous Ca2+ oscillations leading to oocytes with prolonged low CSF level and high APC level, which resulted in defective nuclear envelope breakdown and DNA replication. We have identified an RGS12 variant as the potential cause of female infertility characterized by arrest at the PN stage during multiple IVF and ICSI.
Collapse
Affiliation(s)
- Tianzhong Ma
- Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Songxia Zhou
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Xuezhen Xie
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jingyao Chen
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jing Wang
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Guohong Zhang
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
10
|
Iglesias-Romero AB, Soto T, Flor-Parra I, Salas-Pino S, Ruiz-Romero G, Gould KL, Cansado J, Daga RR. MAPK-dependent control of mitotic progression in S. pombe. BMC Biol 2024; 22:71. [PMID: 38523261 PMCID: PMC10962199 DOI: 10.1186/s12915-024-01865-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 03/08/2024] [Indexed: 03/26/2024] Open
Abstract
BACKGROUND Mitogen-activated protein kinases (MAPKs) preserve cell homeostasis by transducing physicochemical fluctuations of the environment into multiple adaptive responses. These responses involve transcriptional rewiring and the regulation of cell cycle transitions, among others. However, how stress conditions impinge mitotic progression is largely unknown. The mitotic checkpoint is a surveillance mechanism that inhibits mitotic exit in situations of defective chromosome capture, thus preventing the generation of aneuploidies. In this study, we investigate the role of MAPK Pmk1 in the regulation of mitotic exit upon stress. RESULTS We show that Schizosaccharomyces pombe cells lacking Pmk1, the MAP kinase effector of the cell integrity pathway (CIP), are hypersensitive to microtubule damage and defective in maintaining a metaphase arrest. Epistasis analysis suggests that Pmk1 is involved in maintaining spindle assembly checkpoint (SAC) signaling, and its deletion is additive to the lack of core SAC components such as Mad2 and Mad3. Strikingly, pmk1Δ cells show up to twofold increased levels of the anaphase-promoting complex (APC/C) activator Cdc20Slp1 during unperturbed growth. We demonstrate that Pmk1 physically interacts with Cdc20Slp1 N-terminus through a canonical MAPK docking site. Most important, the Cdc20Slp1 pool is rapidly degraded in stressed cells undergoing mitosis through a mechanism that requires MAPK activity, Mad3, and the proteasome, thus resulting in a delayed mitotic exit. CONCLUSIONS Our data reveal a novel function of MAPK in preventing mitotic exit and activation of cytokinesis in response to stress. The regulation of Cdc20Slp1 turnover by MAPK Pmk1 provides a key mechanism by which the timing of mitotic exit can be adjusted relative to environmental conditions.
Collapse
Affiliation(s)
| | - Terersa Soto
- Yeast Physiology Group, Department of Genetics and Microbiology, Facultad de Biología, Universidad de Murcia, Murcia, 30071, Spain
| | - Ignacio Flor-Parra
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Seville, 41013, Spain
| | - Silvia Salas-Pino
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Seville, 41013, Spain
| | - Gabriel Ruiz-Romero
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Seville, 41013, Spain
| | - Kathleen L Gould
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37240, USA
| | - José Cansado
- Yeast Physiology Group, Department of Genetics and Microbiology, Facultad de Biología, Universidad de Murcia, Murcia, 30071, Spain.
| | - Rafael R Daga
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Seville, 41013, Spain.
| |
Collapse
|
11
|
Tu Y, Zhang H, Xia J, Zhao Y, Yang R, Feng J, Ma X, Li J. SETDB2 interacts with BUBR1 to induce accurate chromosome segregation independently of its histone methyltransferase activity. FEBS Open Bio 2024; 14:444-454. [PMID: 38151757 PMCID: PMC10909981 DOI: 10.1002/2211-5463.13761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/03/2023] [Accepted: 12/27/2023] [Indexed: 12/29/2023] Open
Abstract
SETDB2 is a H3K9 histone methyltransferase required for accurate chromosome segregation. Its H3K9 histone methyltransferase activity was reported to be associated with chromosomes during metaphase. Here, we confirm that SETDB2 is required for mitosis and accurate chromosome segregation. However, these functions are independent of its histone methyltransferase activity. Further analysis showed that SETDB2 can interact with BUBR1, and is required for CDC20 binding to BUBR1 and APC/C complex and CYCLIN B1 degradation. The ability of SETDB2 to regulate the binding of CDC20 to BUBR1 or APC/C complex, and stabilization of CYCLIN B1 are also independent of its histone methyltransferase activity. These results suggest that SETDB2 interacts with BUBR1 to promote binding of CDC20 to BUBR1 and APC3, then degrades CYCLIN B1 to ensure accurate chromosome segregation and mitosis, independently of its histone methyltransferase activity.
Collapse
Affiliation(s)
- Yanhong Tu
- School of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouChina
- The Second Affiliated HospitalThe Chinese University of Hong KongShenzhenChina
| | - Haomiao Zhang
- The Third School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Jialin Xia
- School of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouChina
| | - Yu Zhao
- Anhui University of Science and Technology Affiliated Fengxian HospitalShanghaiChina
| | - Ruifang Yang
- Anhui University of Science and Technology Affiliated Fengxian HospitalShanghaiChina
| | - Jing Feng
- School of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouChina
- The Second Affiliated HospitalThe Chinese University of Hong KongShenzhenChina
- Anhui University of Science and Technology Affiliated Fengxian HospitalShanghaiChina
| | - Xueyun Ma
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Jing Li
- School of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouChina
- Anhui University of Science and Technology Affiliated Fengxian HospitalShanghaiChina
| |
Collapse
|
12
|
Zhang Y, Young R, Garvanska DH, Song C, Zhai Y, Wang Y, Jiang H, Fang J, Nilsson J, Alfieri C, Zhang G. Functional analysis of Cdc20 reveals a critical role of CRY box in mitotic checkpoint signaling. Commun Biol 2024; 7:164. [PMID: 38337031 PMCID: PMC10858191 DOI: 10.1038/s42003-024-05859-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Accurate mitosis is coordinated by the spindle assembly checkpoint (SAC) through the mitotic checkpoint complex (MCC), which inhibits the anaphase-promoting complex or cyclosome (APC/C). As an essential regulator, Cdc20 promotes mitotic exit through activating APC/C and monitors kinetochore-microtubule attachment through activating SAC. Cdc20 requires multiple interactions with APC/C and MCC subunits to elicit these functions. Functionally assessing these interactions within cells requires efficient depletion of endogenous Cdc20, which is highly difficult to achieve by RNA interference (RNAi). Here we generated Cdc20 RNAi-sensitive cell lines which display a penetrant metaphase arrest by a single RNAi treatment. In this null background, we accurately measured the contribution of each known motif of Cdc20 on APC/C and SAC activation. The CRY box, a previously identified degron, was found critical for SAC by promoting MCC formation and its interaction with APC/C. These data reveal additional regulation within the SAC and establish a novel method to interrogate Cdc20.
Collapse
Affiliation(s)
- Yuqing Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Rose Young
- Chester Beatty Laboratories, Structural Biology Division, Institute of Cancer Research, London, UK
| | | | - Chunlin Song
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yujing Zhai
- School of Public Health, Qingdao University, Qingdao, China
| | - Ying Wang
- School of Public Health, Qingdao University, Qingdao, China
| | - Hongfei Jiang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jing Fang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jakob Nilsson
- The NNF Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Claudio Alfieri
- Chester Beatty Laboratories, Structural Biology Division, Institute of Cancer Research, London, UK.
| | - Gang Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.
| |
Collapse
|
13
|
Liu M, Yang S, Yang J, Feng P, Luo F, Zhang Q, Yang L, Jiang H. BubR1 controls starvation-induced lipolysis via IMD signaling pathway in Drosophila. Aging (Albany NY) 2024; 16:3257-3279. [PMID: 38334966 PMCID: PMC10929803 DOI: 10.18632/aging.205533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/08/2024] [Indexed: 02/10/2024]
Abstract
Lipolysis, the key process releasing fat acids to generate energy in adipose tissues, correlates with starvation resistance. Nevertheless, its detail mechanisms remain elusive. BubR1, an essential mitotic regulator, ensures proper chromosome alignment and segregation during mitosis, but its physiological functions are largely unknown. Here, we use Drosophila adult fat body, the major lipid storage organ, to study the functions of BubR1 in lipolysis. We show that both whole body- and fat body-specific BubR1 depletions increase lipid degradation and shorten the lifespan under fasting but not feeding. Relish, the conserved regulator of IMD signaling pathway, acts as the downstream target of BubR1 to control the expression level of Bmm and modulate the lipolysis upon fasting. Thus, our study reveals new functions of BubR1 in starvation-induced lipolysis and provides new insights into the molecular mechanisms of lipolysis mediated by IMD signaling pathway.
Collapse
Affiliation(s)
- Mengyou Liu
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shengye Yang
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingsi Yang
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ping Feng
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Feng Luo
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiaoqiao Zhang
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Li Yang
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hao Jiang
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
14
|
Sun Y, Chen Z, Liu G, Chen X, Shi Z, Feng H, Yu L, Li G, Ding K, Huang H, Zhang Z, Xu S. Discovery of a potent and selective covalent threonine tyrosine kinase (TTK) inhibitor. Bioorg Chem 2024; 143:107053. [PMID: 38159497 DOI: 10.1016/j.bioorg.2023.107053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/07/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Threonine tyrosine kinase (TTK) is a critical component of the spindle assembly checkpoint and plays a pivotal role in mitosis. TTK has been identified as a potential therapeutic target for human cancers. Here, we describe our design, synthesis and evaluation of a class of covalent TTK inhibitors, exemplified by 16 (SYL1073). Compound 16 potently inhibits TTK kinase with an IC50 of 0.016 μM and displays improved selectivity in a panel of kinases. Mass spectrometry analysis reveals that 16 covalently binds to the C604 cysteine residue in the hinge region of the TTK kinase domain. Furthermore, 16 achieves strong potency in inhibiting the growth of various human cancer cell lines, outperforming its relative reversible inhibitor, and eliciting robust downstream effects. Taken together, compound 16 provides a valuable lead compound for further optimization toward the development of drug for treatment of human cancers.
Collapse
Affiliation(s)
- Yaoliang Sun
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhiwen Chen
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Guobin Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaoai Chen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zihan Shi
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Science, 19 Yuquan Road, Beijing 100049, China
| | - Huixu Feng
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lei Yu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Guodong Li
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - He Huang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Zhang Zhang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Shilin Xu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; University of Chinese Academy of Science, 19 Yuquan Road, Beijing 100049, China.
| |
Collapse
|
15
|
Ballmer D, Lou HJ, Ishii M, Turk BE, Akiyoshi B. An unconventional regulatory circuitry involving Aurora B controls anaphase onset and error-free chromosome segregation in trypanosomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.20.576407. [PMID: 38293145 PMCID: PMC10827227 DOI: 10.1101/2024.01.20.576407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Accurate chromosome segregation during mitosis requires that all chromosomes establish stable bi-oriented attachments with the spindle apparatus. Kinetochores form the interface between chromosomes and spindle microtubules and as such are under tight control by complex regulatory circuitry. As part of the chromosomal passenger complex (CPC), the Aurora B kinase plays a central role within this circuitry by destabilizing improper kinetochore-microtubule attachments and relaying the attachment status to the spindle assembly checkpoint, a feedback control system that delays the onset of anaphase by inhibiting the anaphase-promoting complex/cyclosome. Intriguingly, Aurora B is conserved even in kinetoplastids, an evolutionarily divergent group of eukaryotes, whose kinetochores are composed of a unique set of structural and regulatory proteins. Kinetoplastids do not have a canonical spindle checkpoint and it remains unclear how their kinetochores are regulated to ensure the fidelity and timing of chromosome segregation. Here, we show in Trypanosoma brucei, the kinetoplastid parasite that causes African sleeping sickness, that inhibition of Aurora B using an analogue-sensitive approach arrests cells in metaphase, with a reduction in properly bi-oriented kinetochores. Aurora B phosphorylates several kinetochore proteins in vitro, including the N-terminal region of the divergent Bub1-like protein KKT14. Depletion of KKT14 partially overrides the cell cycle arrest caused by Aurora B inhibition, while overexpression of a non-phosphorylatable KKT14 protein results in a prominent delay in the metaphase-to-anaphase transition. Finally, we demonstrate using a nanobody-based system that re-targeting the catalytic module of the CPC to the outer kinetochore is sufficient to promote mitotic exit but causes massive chromosome mis-segregation in anaphase. Our results indicate that the CPC and KKT14 are involved in an unconventional pathway controlling mitotic exit and error-free chromosome segregation in trypanosomes.
Collapse
Affiliation(s)
- Daniel Ballmer
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, United Kingdom
| | - Hua Jane Lou
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Midori Ishii
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, United Kingdom
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Max Born Crescent Edinburgh, EH9 3BF, United Kingdom
| | - Benjamin E. Turk
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Bungo Akiyoshi
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, United Kingdom
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Max Born Crescent Edinburgh, EH9 3BF, United Kingdom
| |
Collapse
|
16
|
Zhao L, Ye S, Jing S, Gao YJ, He T. Targeting TRIP13 for overcoming anticancer drug resistance (Review). Oncol Rep 2023; 50:202. [PMID: 37800638 PMCID: PMC10565899 DOI: 10.3892/or.2023.8639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/30/2023] [Indexed: 10/07/2023] Open
Abstract
Cancer is one of the greatest dangers to human wellbeing and survival. A key barrier to effective cancer therapy is development of resistance to anti‑cancer medications. In cancer cells, the AAA+ ATPase family member thyroid hormone receptor interactor 13 (TRIP13) is key in promoting treatment resistance. Nonetheless, knowledge of the molecular processes underlying TRIP13‑based resistance to anticancer therapies is lacking. The present study evaluated the function of TRIP13 expression in anticancer drug resistance and potential methods to overcome this resistance. Additionally, the underlying mechanisms by which TRIP13 promotes resistance to anticancer drugs were explored, including induction of mitotic checkpoint complex surveillance system malfunction, promotion of DNA repair, the enhancement of autophagy and the prevention of immunological clearance. The effects of combination treatment, which include a TRIP13 inhibitor in addition to other inhibitors, were discussed. The present study evaluated the literature on TRIP13 as a possible target and its association with anticancer drug resistance, which may facilitate improvements in current anticancer therapeutic options.
Collapse
Affiliation(s)
- Liwen Zhao
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, P.R. China
| | - Siyu Ye
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, P.R. China
| | - Shengnan Jing
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, P.R. China
| | - Yong-Jing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, P.R. China
| | - Tianzhen He
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, P.R. China
| |
Collapse
|
17
|
Jiang W, Yang X, Shi K, Zhang Y, Shi X, Wang J, Wang Y, Chenyan A, Shan J, Wang Y, Chang J, Chen R, Zhou T, Zhu Y, Yu Y, Li C, Li X. MAD2 activates IGF1R/PI3K/AKT pathway and promotes cholangiocarcinoma progression by interfering USP44/LIMA1 complex. Oncogene 2023; 42:3344-3357. [PMID: 37752233 DOI: 10.1038/s41388-023-02849-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 09/28/2023]
Abstract
Spindle assembly checkpoint (SAC) plays an essential part in facilitating normal cell division. However, the clinicopathological and biological significance of mitotic arrest deficient 2 like 1 (MAD2/MAD2L1), a highly conserved member of SAC in cholangiocarcinoma (CCA) remain unclear. We aim to determine the role and mechanism of MAD2 in CCA progression. In the study, we found up-regulated MAD2 facilitated CCA progression and induced lymphatic metastasis dependent on USP44/LIMA1/PI3K/AKT pathway. MAD2 interfered the binding of USP44 to LIMA1 by sequestrating more USP44 in nuclei, causing impaired formation of USP44/LIMA1 complex and enhanced LIMA1 K48 (Lys48)-linked ubiquitination. In therapeutic perspective, the data combined eleven cases of CCA PDTX model showed that high-MAD2 inhibits tumor necrosis and diminishes the inhibition of cell viability after treated with gemcitabine-based regimens. Immunohistochemistry (IHC) analysis of tissue microarray (TMA) for CCA patients revealed that high-MAD2, low-USP44 or low-LIMA1 level are correlated with worse survival for patients. Together, MAD2 activates PI3K/AKT pathway, promotes cancer progression and induces gemcitabine chemo-resistance in CCA. These findings suggest that MAD2 might be an excellent indicator in prognosis analysis and chemotherapy guidance for CCA patients.
Collapse
Affiliation(s)
- Wangjie Jiang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu, China
| | - Xiao Yang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kuangheng Shi
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yaodong Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu, China
| | - Xiaoli Shi
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jifei Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuming Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Anlan Chenyan
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jijun Shan
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yirui Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiang Chang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ruixiang Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanping Zhu
- Personaloncology Biological Technology Co., Ltd, Nanjing, Jiangsu, China
| | - Yue Yu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu, China
| | - Changxian Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu, China.
| | - Xiangcheng Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu, China.
| |
Collapse
|
18
|
Kim JH, Patel R. Mad2B forms a complex with Cdc20, Cdc27, Rev3 and Rev1 in response to cisplatin-induced DNA damage. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2023; 27:427-436. [PMID: 37641805 PMCID: PMC10466067 DOI: 10.4196/kjpp.2023.27.5.427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 04/18/2023] [Accepted: 07/18/2023] [Indexed: 08/31/2023]
Abstract
Mitotic arrest deficient 2 like 2 (Mad2L2, also known as Mad2B), the human homologue of the yeast Rev7 protein, is a regulatory subunit of DNA polymerase ζ that shares high sequence homology with Mad2, the mitotic checkpoint protein. Previously, we demonstrated the involvement of Mad2B in the cisplatin-induced DNA damage response. In this study, we extend our findings to show that Mad2B is recruited to sites of DNA damage in human cancer cells in response to cisplatin treatment. We found that in undamaged cells, Mad2B exists in a complex with Polζ-Rev1 and the APC/C subunit Cdc27. Following cisplatin-induced DNA damage, we observed an increase in the recruitment of Mad2B and Cdc20 (the activators of the APC/C), to the complex. The involvement of Mad2B-Cdc20-APC/C during DNA damage has not been reported before and suggests that the APC/C is activated following cisplatin-induced DNA damage. Using an in vitro ubiquitination assay, our data confirmed Mad2B-dependent activation of APC/C in cisplatin-treated cells. Mad2B may act as an accelerator for APC/C activation during DNA damage response. Our data strongly suggest a role for Mad2B-APC/C-Cdc20 in the ubiquitination of proteins involved in the DNA damage response.
Collapse
Affiliation(s)
- Ju Hwan Kim
- Department of Pharmacology, College of Medicine, Dankook University, Cheonan 31116, Korea
| | - Rajnikant Patel
- Department of Molecular and Cell Biology, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
19
|
Ide AH, DeLuca KF, Wiggan O, Markus SM, DeLuca JG. The role of kinetochore dynein in checkpoint silencing is restricted to disassembly of the corona. Mol Biol Cell 2023; 34:ar76. [PMID: 37126397 PMCID: PMC10295480 DOI: 10.1091/mbc.e23-04-0130] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 05/02/2023] Open
Abstract
During mitosis, kinetochore-microtubule attachments are monitored by a molecular surveillance system known as the spindle assembly checkpoint. The prevailing model posits that dynein evicts checkpoint proteins (e.g., Mad1, Mad2) from stably attached kinetochores by transporting them away from kinetochores, thus contributing to checkpoint silencing. However, the mechanism by which dynein performs this function, and its precise role in checkpoint silencing remain unresolved. Here, we find that dynein's role in checkpoint silencing is restricted to evicting checkpoint effectors from the fibrous corona, and not the outer kinetochore. Dynein evicts these molecules from the corona in a manner that does not require stable, end-on microtubule attachments. Thus, by disassembling the corona through indiscriminate microtubule encounters, dynein primes the checkpoint signaling apparatus so it can respond to stable end-on microtubule attachments and permit cells to progress through mitosis. Accordingly, we find that dynein function in checkpoint silencing becomes largely dispensable in cells in which checkpoint effectors are excluded from the corona.
Collapse
Affiliation(s)
- Amy H. Ide
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523
| | - Keith F. DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523
| | - O’Neil Wiggan
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523
| | - Steven M. Markus
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523
| | - Jennifer G. DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523
| |
Collapse
|
20
|
Hu S, Jiang C, Gao M, Zhang D, Yao N, Zhang J, Jin Q. Discovery of pyrazolo[3,4-b]pyridine derivatives as novel and potent Mps1 inhibitors for the treatment of cancer. Eur J Med Chem 2023; 253:115334. [PMID: 37037136 DOI: 10.1016/j.ejmech.2023.115334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/20/2023] [Accepted: 03/30/2023] [Indexed: 04/12/2023]
Abstract
Monopolar spindle kinase 1 (Mps1) is a key element of the mitotic checkpoint and clinically evaluated as a target in the treatment of aggressive tumors. With this aim, a set of pyrazolo[3,4-b]pyridine-based compounds as new Mps1 inhibitors was investigated through a multidisciplinary approach, based on virtual screening, chemical synthesis and biological evaluation. One of the representative compounds, 31, exhibited strong kinase inhibitory potency against Mps1 with an IC50 value of 2.596 nM and significantly inhibited proliferation of cancer cells, especially MDA-MB-468 and MV4-11 cells. Compound 31 also displayed reasonable kinome selectivity against a panel of 606 wild-type kinases at 1 μM. Moreover, compound 31 exhibited suitable preclinical pharmacokinetic parameters and a promising pharmacodynamic profile. Further, compound 31 showed good antitumor efficacy in MDA-MB-468 xenograft model with no obvious toxicity. Overall, compound 31 was identified as a potential Mps1 inhibitor for cancer therapy and deserve further research.
Collapse
Affiliation(s)
- Shihe Hu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China; SkyRun Pharma Co., Ltd., No. 9 Weidi Road, Nanjing, 210046, PR China
| | - Cuihua Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Meng Gao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Dongjian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Nan Yao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Jian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China.
| | - Qiaomei Jin
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China.
| |
Collapse
|
21
|
MacKenzie A, Vicory V, Lacefield S. Meiotic cells escape prolonged spindle checkpoint activity through kinetochore silencing and slippage. PLoS Genet 2023; 19:e1010707. [PMID: 37018287 PMCID: PMC10109492 DOI: 10.1371/journal.pgen.1010707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/17/2023] [Accepted: 03/20/2023] [Indexed: 04/06/2023] Open
Abstract
To prevent chromosome mis-segregation, a surveillance mechanism known as the spindle checkpoint delays the cell cycle if kinetochores are not attached to spindle microtubules, allowing the cell additional time to correct improper attachments. During spindle checkpoint activation, checkpoint proteins bind the unattached kinetochore and send a diffusible signal to inhibit the anaphase promoting complex/cyclosome (APC/C). Previous work has shown that mitotic cells with depolymerized microtubules can escape prolonged spindle checkpoint activation in a process called mitotic slippage. During slippage, spindle checkpoint proteins bind unattached kinetochores, but the cells cannot maintain the checkpoint arrest. We asked if meiotic cells had as robust of a spindle checkpoint response as mitotic cells and whether they also undergo slippage after prolonged spindle checkpoint activity. We performed a direct comparison between mitotic and meiotic budding yeast cells that signal the spindle checkpoint through two different assays. We find that the spindle checkpoint delay is shorter in meiosis I or meiosis II compared to mitosis, overcoming a checkpoint arrest approximately 150 minutes earlier in meiosis than in mitosis. In addition, cells in meiosis I escape spindle checkpoint signaling using two mechanisms, silencing the checkpoint at the kinetochore and through slippage. We propose that meiotic cells undertake developmentally-regulated mechanisms to prevent persistent spindle checkpoint activity to ensure the production of gametes.
Collapse
Affiliation(s)
- Anne MacKenzie
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
| | - Victoria Vicory
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
| | - Soni Lacefield
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
- Department of Biochemistry and Cell Biology, the Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| |
Collapse
|
22
|
Chen C, Piano V, Alex A, Han SJY, Huis In 't Veld PJ, Roy B, Fergle D, Musacchio A, Joglekar AP. The structural flexibility of MAD1 facilitates the assembly of the Mitotic Checkpoint Complex. Nat Commun 2023; 14:1529. [PMID: 36934097 PMCID: PMC10024682 DOI: 10.1038/s41467-023-37235-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 03/08/2023] [Indexed: 03/20/2023] Open
Abstract
The spindle assembly checkpoint (SAC) safeguards the genome during cell division by generating an effector molecule known as the Mitotic Checkpoint Complex (MCC). The MCC comprises two subcomplexes: BUBR1:BUB3 and CDC20:MAD2, and the formation of CDC20:MAD2 is the rate-limiting step during MCC assembly. Recent studies show that the rate of CDC20:MAD2 formation is significantly accelerated by the cooperative binding of CDC20 to the SAC proteins MAD1 and BUB1. However, the molecular basis for this acceleration is not fully understood. Here, we demonstrate that the structural flexibility of MAD1 at a conserved hinge near the C-terminus is essential for catalytic MCC assembly. This MAD1 hinge enables the MAD1:MAD2 complex to assume a folded conformation in vivo. Importantly, truncating the hinge reduces the rate of MCC assembly in vitro and SAC signaling in vivo. Conversely, mutations that preserve hinge flexibility retain SAC signaling, indicating that the structural flexibility of the hinge, rather than a specific amino acid sequence, is important for SAC signaling. We summarize these observations as the 'knitting model' that explains how the folded conformation of MAD1:MAD2 promotes CDC20:MAD2 assembly.
Collapse
Affiliation(s)
- Chu Chen
- Biophysics, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne, 50931, Germany
| | - Valentina Piano
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, 44227, Germany
- Institute of Human Genetics, University Hospital Cologne, Cologne, 50931, Germany
| | - Amal Alex
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, 44227, Germany
| | - Simon J Y Han
- Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Pim J Huis In 't Veld
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, 44227, Germany
| | - Babhrubahan Roy
- Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Daniel Fergle
- Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Andrea Musacchio
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, 44227, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, 45141, Germany
| | - Ajit P Joglekar
- Biophysics, University of Michigan, Ann Arbor, MI, 48109, USA.
- Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
23
|
Comprehensive analysis of BUBs gene family in lung adenocarcinoma with immunological analysis. Aging (Albany NY) 2023; 15:810-829. [PMID: 36787437 PMCID: PMC9970319 DOI: 10.18632/aging.204517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/05/2023] [Indexed: 02/16/2023]
Abstract
Lung adenocarcinoma (LUAD) is one of the most commonly malignant tumors, and major challenges remain in the treatment of LUAD. Budding uninhibited by benzimidazole 1/3 (BUB1/3) play significant roles in the process of spindle-assembly checkpoint (SAC) during mitosis. However, their roles in LUAD have not been established. Here, we performed an immunological analysis of BUB1/3 in LUAD using a comprehensive bioinformatics approach, quantitative real-time-PCR and Western blotting technique. Our results indicated that the expression levels of BUB1 and BUB3 in LUAD samples were higher than the expression levels in the control groups and were associated with some clinicopathologic parameters in patients with LUAD. BUB1/3 and their related genes were enriched in cell immune, and the immune infiltration analysis revealed that the BUB1/3 expression profile was significantly correlated with characteristics of immune cell infiltration. Survival analysis showed that the disease-free survival and overall survival of patients with LUAD decreased with an increase in the BUB1/3 expression levels. The mRNA and protein expression levels of BUB1 and BUB3 in each of the LUAD cell lines were upregulated to varying degrees. BUB1 and BUB3 are the potential immunological therapeutic intervention targets for patients with LUAD.
Collapse
|
24
|
Abdelbaki A, Ascanelli C, Okoye CN, Akman HB, Janson G, Min M, Marcozzi C, Hagting A, Grant R, De Luca M, Asteriti IA, Guarguaglini G, Paiardini A, Lindon C. Revisiting degron motifs in human AURKA required for its targeting by APC/C FZR1. Life Sci Alliance 2023; 6:6/2/e202201372. [PMID: 36450448 PMCID: PMC9713472 DOI: 10.26508/lsa.202201372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 12/02/2022] Open
Abstract
Mitotic kinase Aurora A (AURKA) diverges from other kinases in its multiple active conformations that may explain its interphase roles and the limited efficacy of drugs targeting the kinase pocket. Regulation of AURKA activity by the cell is critically dependent on destruction mediated by the anaphase-promoting complex (APC/CFZR1) during mitotic exit and G1 phase and requires an atypical N-terminal degron in AURKA called the "A-box" in addition to a reported canonical D-box degron in the C-terminus. Here, we find that the reported C-terminal D-box of AURKA does not act as a degron and instead mediates essential structural features of the protein. In living cells, the N-terminal intrinsically disordered region of AURKA containing the A-box is sufficient to confer FZR1-dependent mitotic degradation. Both in silico and in cellulo assays predict the QRVL short linear interacting motif of the A-box to be a phospho-regulated D-box. We propose that degradation of full-length AURKA also depends on an intact C-terminal domain because of critical conformational parameters permissive for both activity and mitotic degradation of AURKA.
Collapse
Affiliation(s)
- Ahmed Abdelbaki
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | - Cynthia N Okoye
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - H Begum Akman
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Giacomo Janson
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Mingwei Min
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Chiara Marcozzi
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Anja Hagting
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Rhys Grant
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Maria De Luca
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Italia Anna Asteriti
- Institute of Molecular Biology and Pathology, National Research Council of Italy, c/o Sapienza University of Rome, Rome, Italy
| | - Giulia Guarguaglini
- Institute of Molecular Biology and Pathology, National Research Council of Italy, c/o Sapienza University of Rome, Rome, Italy
| | | | - Catherine Lindon
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
25
|
MacKenzie A, Vicory V, Lacefield S. Meiotic Cells Escape Prolonged Spindle Checkpoint Activity Through Premature Silencing and Slippage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.02.522494. [PMID: 36711621 PMCID: PMC9881877 DOI: 10.1101/2023.01.02.522494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
To prevent chromosome mis-segregation, a surveillance mechanism known as the spindle checkpoint delays the cell cycle if kinetochores are not attached to spindle microtubules, allowing the cell additional time to correct improper attachments. During spindle checkpoint activation, checkpoint proteins bind the unattached kinetochore and send a diffusible signal to inhibit the anaphase promoting complex/cyclosome (APC/C). Previous work has shown that mitotic cells with depolymerized microtubules can escape prolonged spindle checkpoint activation in a process called mitotic slippage. During slippage, spindle checkpoint proteins bind unattached kinetochores, but the cells cannot maintain the checkpoint arrest. We asked if meiotic cells had as robust of a spindle checkpoint response as mitotic cells and whether they also undergo slippage after prolonged spindle checkpoint activity. We performed a direct comparison between mitotic and meiotic budding yeast cells that signal the spindle checkpoint due to a lack of either kinetochore-microtubule attachments or due to a loss of tension-bearing attachments. We find that the spindle checkpoint is not as robust in meiosis I or meiosis II compared to mitosis, overcoming a checkpoint arrest approximately 150 minutes earlier in meiosis. In addition, cells in meiosis I escape spindle checkpoint signaling using two mechanisms, silencing the checkpoint at the kinetochore and through slippage. We propose that meiotic cells undertake developmentally-regulated mechanisms to prevent persistent spindle checkpoint activity to ensure the production of gametes. AUTHOR SUMMARY Mitosis and meiosis are the two major types of cell divisions. Mitosis gives rise to genetically identical daughter cells, while meiosis is a reductional division that gives rise to gametes. Cell cycle checkpoints are highly regulated surveillance mechanisms that prevent cell cycle progression when circumstances are unfavorable. The spindle checkpoint promotes faithful chromosome segregation to safeguard against aneuploidy, in which cells have too many or too few chromosomes. The spindle checkpoint is activated at the kinetochore and then diffuses to inhibit cell cycle progression. Although the checkpoint is active in both mitosis and meiosis, most studies involving checkpoint regulation have been performed in mitosis. By activating the spindle checkpoint in both mitosis and meiosis in budding yeast, we show that cells in meiosis elicit a less persistent checkpoint signal compared to cells in mitosis. Further, we show that cells use distinct mechanisms to escape the checkpoint in mitosis and meiosis I. While cells in mitosis and meiosis II undergo anaphase onset while retaining checkpoint proteins at the kinetochore, cells in meiosis I prematurely lose checkpoint protein localization at the kinetochore. If the mechanism to remove the checkpoint components from the kinetochore is disrupted, meiosis I cells can still escape checkpoint activity. Together, these results highlight that cell cycle checkpoints are differentially regulated during meiosis to avoid long delays and to allow gametogenesis.
Collapse
Affiliation(s)
- Anne MacKenzie
- Department of Biology, Indiana University, Bloomington, IN USA
| | - Victoria Vicory
- Department of Biology, Indiana University, Bloomington, IN USA
| | - Soni Lacefield
- Department of Biology, Indiana University, Bloomington, IN USA,Department of Biochemistry and Cell Biology, the Geisel School of Medicine at Dartmouth, Hanover, NH USA,To whom correspondence should be addressed to Soni Lacefield:
| |
Collapse
|
26
|
Bhattacharjee D, Kaveti S, Jain N. APC/C CDH1 ubiquitinates STAT3 in mitosis. Int J Biochem Cell Biol 2023; 154:106333. [PMID: 36400381 DOI: 10.1016/j.biocel.2022.106333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
STAT3, an oncogene drives tumor growth and is associated with poor prognosis. However, small molecule-based STAT3 inhibitors were unsuccessful in clinics. Recently, STAT3 degraders that ubiquitinate STAT3 were found to elicit long-lasting anti-tumor responses. Thus, triggering STAT3 ubiquitination in cancers is a better strategy than STAT3 inhibition. However, not much is known about the identity of E3-ligases that ubiquitinate STAT3 in cancers. Therefore, to design better therapies to degrade STAT3, we sought to identify E3-ligases that ubiquitinate STAT3 in cancer cells. To answer this question, we determined the cell cycle-dependent ubiquitination of STAT3 in HEK293T cells and examined the link between STAT3 dephosphorylation and ubiquitination. We found that STAT3 is more strongly ubiquitinated in mitosis than in other phases of the cell cycle. We observed that APC/C CDH1 binds and ubiquitinates STAT3 in mitosis. Further, we also found that inhibiting phosphatases decreases STAT3 ubiquitination. We conclude that APC/C CDH1 ubiquitinates STAT3 in mitosis. We suggest that mitosis can be a potential therapeutic window for treating STAT3-activated cancers.
Collapse
Affiliation(s)
- Debanjan Bhattacharjee
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Sector 19, Kamala Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India
| | - Sreeram Kaveti
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Sector 19, Kamala Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India
| | - Nishant Jain
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Sector 19, Kamala Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India.
| |
Collapse
|
27
|
Patra D, Bhavya K, Ramprasad P, Kalia M, Pal D. Anti-cancer drug molecules targeting cancer cell cycle and proliferation. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 135:343-395. [PMID: 37061337 DOI: 10.1016/bs.apcsb.2022.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Cancer, a vicious clinical burden that potentiates maximum fatality for humankind, arises due to unregulated excessive cell division and proliferation through an eccentric expression of cell cycle regulator proteins. A set of evolutionarily conserved machinery controls the cell cycle in an extremely precise manner so that a cell that went through the cycle can produce a genetically identical copy. To achieve perfection, several checkpoints were placed in the cycle for surveillance; so, errors during the division were rectified by the repair strategies. However, irreparable damage leads to exit from the cell cycle and induces programmed cell death. In comparison to a normal cell, cancer cells facilitate the constitutive activation of many dormant proteins and impede negative regulators of the checkpoint. Extensive studies in the last few decades on cell division and proliferation of cancer cells elucidate the molecular mechanism of the cell-cycle regulators that are often targeted for the development of anti-cancer therapy. Each phase of the cell cycle has been regulated by a unique set of proteins including master regulators Cyclins, and CDKs, along with the accessory proteins such as CKI, Cdc25, error-responsive proteins, and various kinase proteins mainly WEE1 kinases, Polo-like kinases, and Aurora kinases that control cell division. Here in this chapter, we have analytically discussed the role of cell cycle regulators and proliferation factors in cancer progression and the rationale of using various cell cycle-targeting drug molecules as anti-cancer therapy.
Collapse
Affiliation(s)
- Debarun Patra
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Kumari Bhavya
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Palla Ramprasad
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Moyna Kalia
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Durba Pal
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India.
| |
Collapse
|
28
|
Fischer ES. Kinetochore‐catalyzed MCC
formation: A structural perspective. IUBMB Life 2022; 75:289-310. [PMID: 36518060 DOI: 10.1002/iub.2697] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/08/2022] [Indexed: 12/23/2022]
Abstract
The spindle assembly checkpoint (SAC) is a cellular surveillance mechanism that functions to ensure accurate chromosome segregation during mitosis. Macromolecular complexes known as kinetochores, act as the interface of sister chromatid attachment to spindle microtubules. In response to unattached kinetochores, the SAC activates its effector, the mitotic checkpoint complex (MCC), which delays mitotic exit until all sister chromatid pairs have achieved successful attachment to the bipolar mitotic spindle. Formation of the MCC (composed of Mad2, BubR1, Bub3 and Cdc20) is regulated by an Mps1 kinase-dependent phosphorylation signaling cascade which assembles and repositions components of the MCC onto a catalytic scaffold. This scaffold functions to catalyze the conversion of the HORMA-domain protein Mad2 from an "inactive" open-state (O-Mad2) into an "active" closed-Mad2 (C-Mad2), and simultaneous Cdc20 binding. Here, our current understanding of the molecular mechanisms underlying the kinetic barrier to C-Mad2:Cdc20 formation will be reviewed. Recent progress in elucidating the precise molecular choreography orchestrated by the catalytic scaffold to rapidly assemble the MCC will be examined, and unresolved questions will be highlighted. Ultimately, understanding how the SAC rapidly activates the checkpoint not only provides insights into how cells maintain genomic integrity during mitosis, but also provides a paradigm for how cells can utilize molecular switches, including other HORMA domain-containing proteins, to make rapid changes to a cell's physiological state.
Collapse
Affiliation(s)
- Elyse S. Fischer
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus Cambridge UK
| |
Collapse
|
29
|
Fischer ES, Yu CWH, Hevler JF, McLaughlin SH, Maslen SL, Heck AJR, Freund SMV, Barford D. Juxtaposition of Bub1 and Cdc20 on phosphorylated Mad1 during catalytic mitotic checkpoint complex assembly. Nat Commun 2022; 13:6381. [PMID: 36289199 PMCID: PMC9605988 DOI: 10.1038/s41467-022-34058-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/11/2022] [Indexed: 12/25/2022] Open
Abstract
In response to improper kinetochore-microtubule attachments in mitosis, the spindle assembly checkpoint (SAC) assembles the mitotic checkpoint complex (MCC) to inhibit the anaphase-promoting complex/cyclosome, thereby delaying entry into anaphase. The MCC comprises Mad2:Cdc20:BubR1:Bub3. Its assembly is catalysed by unattached kinetochores on a Mad1:Mad2 platform. Mad1-bound closed-Mad2 (C-Mad2) recruits open-Mad2 (O-Mad2) through self-dimerization. This interaction, combined with Mps1 kinase-mediated phosphorylation of Bub1 and Mad1, accelerates MCC assembly, in a process that requires O-Mad2 to C-Mad2 conversion and concomitant binding of Cdc20. How Mad1 phosphorylation catalyses MCC assembly is poorly understood. Here, we characterized Mps1 phosphorylation of Mad1 and obtained structural insights into a phosphorylation-specific Mad1:Cdc20 interaction. This interaction, together with the Mps1-phosphorylation dependent association of Bub1 and Mad1, generates a tripartite assembly of Bub1 and Cdc20 onto the C-terminal domain of Mad1 (Mad1CTD). We additionally identify flexibility of Mad1:Mad2 that suggests how the Cdc20:Mad1CTD interaction brings the Mad2-interacting motif (MIM) of Cdc20 near O-Mad2. Thus, Mps1-dependent formation of the MCC-assembly scaffold functions to position and orient Cdc20 MIM near O-Mad2, thereby catalysing formation of C-Mad2:Cdc20.
Collapse
Affiliation(s)
- Elyse S Fischer
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge, CB2 0QH, UK.
| | - Conny W H Yu
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Johannes F Hevler
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, 3584 CH, Utrecht, The Netherlands
- Netherlands Proteomics Center, University of Utrecht, 3584 CH, Utrecht, The Netherlands
| | - Stephen H McLaughlin
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Sarah L Maslen
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, 3584 CH, Utrecht, The Netherlands
- Netherlands Proteomics Center, University of Utrecht, 3584 CH, Utrecht, The Netherlands
| | - Stefan M V Freund
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - David Barford
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge, CB2 0QH, UK.
| |
Collapse
|
30
|
Chen OJ, Castellsagué E, Moustafa-Kamal M, Nadaf J, Rivera B, Fahiminiya S, Wang Y, Gamache I, Pacifico C, Jiang L, Carrot-Zhang J, Witkowski L, Berghuis AM, Schönberger S, Schneider D, Hillmer M, Bens S, Siebert R, Stewart CJR, Zhang Z, Chao WCH, Greenwood CMT, Barford D, Tischkowitz M, Majewski J, Foulkes WD, Teodoro JG. Germline Missense Variants in CDC20 Result in Aberrant Mitotic Progression and Familial Cancer. Cancer Res 2022; 82:3499-3515. [PMID: 35913887 DOI: 10.1158/0008-5472.can-21-3956] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 04/12/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022]
Abstract
CDC20 is a coactivator of the anaphase promoting complex/cyclosome (APC/C) and is essential for mitotic progression. APC/CCDC20 is inhibited by the spindle assembly checkpoint (SAC), which prevents premature separation of sister chromatids and aneuploidy in daughter cells. Although overexpression of CDC20 is common in many cancers, oncogenic mutations have never been identified in humans. Using whole-exome sequencing, we identified heterozygous missense CDC20 variants (L151R and N331K) that segregate with ovarian germ cell tumors in two families. Functional characterization showed these mutants retain APC/C activation activity but have impaired binding to BUBR1, a component of the SAC. Expression of L151R and N331K variants promoted mitotic slippage in HeLa cells and primary skin fibroblasts derived from carriers. Generation of mice carrying the N331K variant using CRISPR-Cas9 showed that, although homozygous N331K mice were nonviable, heterozygotes displayed accelerated oncogenicity of Myc-driven cancers. These findings highlight an unappreciated role for CDC20 variants as tumor-promoting genes. SIGNIFICANCE Two germline CDC20 missense variants that segregate with cancer in two families compromise the spindle assembly checkpoint and lead to aberrant mitotic progression, which could predispose cells to transformation. See related commentary by Villarroya-Beltri and Malumbres, p. 3432.
Collapse
Affiliation(s)
- Owen J Chen
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Ester Castellsagué
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Division of Medical Genetics and Cancer Axis, Lady Davis Institute, Segal Cancer Centre, Jewish General Hospital, Montréal, Québec, Canada
- Translational Research Laboratory, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Mohamed Moustafa-Kamal
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Javad Nadaf
- McGill University and Génome Québec Innovation Centre, Montréal, Québec, Canada
| | - Barbara Rivera
- Cancer Axis, Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada
- Hereditary Cancer Programme, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Barcelona, Spain
- Gerald Bronfman Department of Oncology, McGill University, Montréal, Québec, Canada
| | - Somayyeh Fahiminiya
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Yilin Wang
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Isabelle Gamache
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
| | - Caterina Pacifico
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Department of Biology, McGill University, Montréal, Québec, Canada
| | - Lai Jiang
- Cancer Axis, Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada
- Department of Epidemiology, Biostatistics & Occupational Health, McGill University, Montréal, Québec, Canada
| | - Jian Carrot-Zhang
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Leora Witkowski
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Division of Medical Genetics and Cancer Axis, Lady Davis Institute, Segal Cancer Centre, Jewish General Hospital, Montréal, Québec, Canada
| | - Albert M Berghuis
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
- Centre de Recherche en Biologie Structurale, McGill University, Montréal, Québec, Canada
- Department of Microbiology and Immunology, Montréal, Québec, Canada
| | - Stefan Schönberger
- Department of Pediatric Hematology and Oncology, Pediatrics III, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Dominik Schneider
- Clinic of Pediatrics, Dortmund Municipal Hospital, Dortmund, Germany
| | - Morten Hillmer
- Institute of Human Genetics, University of Ulm & Ulm University Medical Center, Ulm, Germany
| | - Susanne Bens
- Institute of Human Genetics, University of Ulm & Ulm University Medical Center, Ulm, Germany
| | - Reiner Siebert
- Institute of Human Genetics, University of Ulm & Ulm University Medical Center, Ulm, Germany
| | - Colin J R Stewart
- Department of Histopathology, King Edward Memorial Hospital, and School for Women's and Infants' Health, University of Western Australia, Perth, Australia
| | - Ziguo Zhang
- Institute of Cancer Research, London, United Kingdom
| | - William C H Chao
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Celia M T Greenwood
- Cancer Axis, Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada
- Department of Epidemiology, Biostatistics & Occupational Health, McGill University, Montréal, Québec, Canada
- Departments of Oncology and Human Genetics, McGill University, Montréal, Québec, Canada
| | - David Barford
- Institute of Cancer Research, London, United Kingdom
| | - Marc Tischkowitz
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Jacek Majewski
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - William D Foulkes
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Division of Medical Genetics and Cancer Axis, Lady Davis Institute, Segal Cancer Centre, Jewish General Hospital, Montréal, Québec, Canada
- Program in Cancer Genetics, Department of Oncology and Human Genetics, McGill University, Montréal, Québec, Canada
- Division of Medical Genetics and Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Jose G Teodoro
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
- Department of Microbiology and Immunology, Montréal, Québec, Canada
| |
Collapse
|
31
|
Bai S, Sun L, Wang X, Wang SM, Luo ZQ, Wang Y, Jin QW. Recovery from spindle checkpoint-mediated arrest requires a novel Dnt1-dependent APC/C activation mechanism. PLoS Genet 2022; 18:e1010397. [PMID: 36108046 PMCID: PMC9514617 DOI: 10.1371/journal.pgen.1010397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/27/2022] [Accepted: 08/24/2022] [Indexed: 11/19/2022] Open
Abstract
The activated spindle assembly checkpoint (SAC) potently inhibits the anaphase-promoting complex/cyclosome (APC/C) to ensure accurate chromosome segregation at anaphase. Early studies have recognized that the SAC should be silenced within minutes to enable rapid APC/C activation and synchronous segregation of chromosomes once all kinetochores are properly attached, but the underlying silencers are still being elucidated. Here, we report that the timely silencing of SAC in fission yeast requires dnt1+, which causes severe thiabendazole (TBZ) sensitivity and increased rate of lagging chromosomes when deleted. The absence of Dnt1 results in prolonged inhibitory binding of mitotic checkpoint complex (MCC) to APC/C and attenuated protein levels of Slp1Cdc20, consequently slows the degradation of cyclin B and securin, and eventually delays anaphase entry in cells released from SAC activation. Interestingly, Dnt1 physically associates with APC/C upon SAC activation. We propose that this association may fend off excessive and prolonged MCC binding to APC/C and help to maintain Slp1Cdc20 stability. This may allow a subset of APC/C to retain activity, which ensures rapid anaphase onset and mitotic exit once SAC is inactivated. Therefore, our study uncovered a new player in dictating the timing and efficacy of APC/C activation, which is actively required for maintaining cell viability upon recovery from the inhibition of APC/C by spindle checkpoint.
Collapse
Affiliation(s)
- Shuang Bai
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Li Sun
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xi Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Shuang-min Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhou-qing Luo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- * E-mail: (ZL); (YW); (QJ)
| | - Yamei Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- * E-mail: (ZL); (YW); (QJ)
| | - Quan-wen Jin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- * E-mail: (ZL); (YW); (QJ)
| |
Collapse
|
32
|
Deng DJ, Wang X, Yue KY, Wang Y, Jin QW. Analysis of the potential role of fission yeast PP2A in spindle assembly checkpoint inactivation. FASEB J 2022; 36:e22524. [PMID: 36006032 DOI: 10.1096/fj.202101884r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 07/26/2022] [Accepted: 08/17/2022] [Indexed: 11/11/2022]
Abstract
As a surveillance mechanism, the activated spindle assembly checkpoint (SAC) potently inhibits the E3 ubiquitin ligase APC/C (anaphase-promoting complex/cyclosome) to ensure accurate chromosome segregation. Although the protein phosphatase 2A (PP2A) has been proposed to be both, directly and indirectly, involved in spindle assembly checkpoint inactivation in mammalian cells, whether it is similarly operating in the fission yeast Schizosaccharomycer pombe has never been demonstrated. Here, we investigated whether fission yeast PP2A is involved in SAC silencing by following the rate of cyclin B (Cdc13) destruction at SPBs during the recovery phase in nda3-KM311 cells released from the inhibition of APC/C by the activated spindle checkpoint. The timing of the SAC inactivation is only slightly delayed when two B56 regulatory subunits (Par1 and Par2) of fission yeast PP2A are absent. Overproduction of individual PP2A subunits either globally in the nda3-KM311 arrest-and-release system or locally in the synthetic spindle checkpoint activation system only slightly suppresses the SAC silencing defects in PP1 deletion (dis2Δ) cells. Our study thus demonstrates that the fission yeast PP2A is not a key regulator actively involved in SAC inactivation.
Collapse
Affiliation(s)
- Da-Jie Deng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Xi Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Kai-Ye Yue
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Yamei Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Quan-Wen Jin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
33
|
Esposito E, Weidemann DE, Rogers JM, Morton CM, Baybay EK, Chen J, Hauf S. Mitotic checkpoint gene expression is tuned by codon usage bias. EMBO J 2022; 41:e107896. [PMID: 35811551 PMCID: PMC9340482 DOI: 10.15252/embj.2021107896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 11/09/2022] Open
Abstract
The mitotic checkpoint (also called spindle assembly checkpoint, SAC) is a signaling pathway that safeguards proper chromosome segregation. Correct functioning of the SAC depends on adequate protein concentrations and appropriate stoichiometries between SAC proteins. Yet very little is known about the regulation of SAC gene expression. Here, we show in the fission yeast Schizosaccharomyces pombe that a combination of short mRNA half-lives and long protein half-lives supports stable SAC protein levels. For the SAC genes mad2+ and mad3+ , their short mRNA half-lives are caused, in part, by a high frequency of nonoptimal codons. In contrast, mad1+ mRNA has a short half-life despite a higher frequency of optimal codons, and despite the lack of known RNA-destabilizing motifs. Hence, different SAC genes employ different strategies of expression. We further show that Mad1 homodimers form co-translationally, which may necessitate a certain codon usage pattern. Taken together, we propose that the codon usage of SAC genes is fine-tuned to ensure proper SAC function. Our work shines light on gene expression features that promote spindle assembly checkpoint function and suggests that synonymous mutations may weaken the checkpoint.
Collapse
Affiliation(s)
- Eric Esposito
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVAUSA
| | - Douglas E Weidemann
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVAUSA
| | - Jessie M Rogers
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVAUSA
| | - Claire M Morton
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVAUSA
| | - Erod Keaton Baybay
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVAUSA
| | - Jing Chen
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVAUSA
| | - Silke Hauf
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVAUSA
| |
Collapse
|
34
|
Bruno S, Ghelli Luserna di Rorà A, Napolitano R, Soverini S, Martinelli G, Simonetti G. CDC20 in and out of mitosis: a prognostic factor and therapeutic target in hematological malignancies. J Exp Clin Cancer Res 2022; 41:159. [PMID: 35490245 PMCID: PMC9055704 DOI: 10.1186/s13046-022-02363-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/11/2022] [Indexed: 12/31/2022] Open
Abstract
Cell division cycle 20 homologue (CDC20) is a well-known regulator of cell cycle, as it controls the correct segregation of chromosomes during mitosis. Many studies have focused on the biological role of CDC20 in cancer development, as alterations of its functionality have been linked to genomic instability and evidence demonstrated that high CDC20 expression levels are associated with poor overall survival in solid cancers. More recently, novel CDC20 functions have been demonstrated or suggested, including the regulation of apoptosis and stemness properties and a correlation with immune cell infiltration. Here, we here summarize and discuss the role of CDC20 inside and outside mitosis, starting from its network of interacting proteins. In the last years, CDC20 has also attracted more interest in the blood cancer field, being overexpressed and showing an association with prognosis both in myeloid and lymphoid malignancies. Preclinical findings showed that selective CDC20 and APC/CCDC20/APC/CCDH1 inhibitors, namely Apcin and proTAME, are effective against lymphoma and multiple myeloma cells, resulting in mitotic arrest and apoptosis and synergizing with clinically-relevant drugs. The evidence and hypothesis presented in this review provide the input for further biological and chemical studies aiming to dissect novel potential CDC20 roles and targeting strategies in hematological malignancies.
Collapse
Affiliation(s)
- Samantha Bruno
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna and Institute of Hematology "L. e A. Seràgnoli", Bologna, Italy
| | - Andrea Ghelli Luserna di Rorà
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", via Piero Maroncelli 40, 47014, Meldola, FC, Italy.
| | - Roberta Napolitano
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Simona Soverini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna and Institute of Hematology "L. e A. Seràgnoli", Bologna, Italy
| | - Giovanni Martinelli
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| | - Giorgia Simonetti
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", via Piero Maroncelli 40, 47014, Meldola, FC, Italy
| |
Collapse
|
35
|
Wang Y, Yu T, Han Y, He Y, Song Y, Guo L, An L, Yang C, Wang F. Phosphorylation of MAD2 at Ser195 Promotes Spindle Checkpoint Defects and Sensitizes Cancer Cells to Radiotherapy in ATM Deficient Cells. Front Cell Dev Biol 2022; 10:817831. [PMID: 35309941 PMCID: PMC8924061 DOI: 10.3389/fcell.2022.817831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/18/2022] [Indexed: 11/13/2022] Open
Abstract
The spindle assembly checkpoint (SAC) is a critical monitoring device in mitosis for the maintenance of genomic stability. Specifically, the SAC complex comprises several factors, including Mad1, Mad2, and Bub1. Ataxia-telangiectasia mutated (ATM) kinase, the crucial regulator in DNA damage response (DDR), also plays a critical role in mitosis by regulating Mad1 dimerization and SAC. Here, we further demonstrated that ATM negatively regulates the phosphorylation of Mad2, another critical component of the SAC, which is also involved in DDR. Mechanistically, we found that phosphorylation of Mad2 is aberrantly increased in ATM-deficient cells. Point-mutation analysis further revealed that Serine 195 mainly mediated Mad2 phosphorylation upon ATM ablation. Functionally, the phosphorylation of Mad2 causes decreased DNA damage repair capacity and is related to the resistance to cancer cell radiotherapy. Altogether, this study unveils the key regulatory role of Mad2 phosphorylation in checkpoint defects and DNA damage repair in ATM-deficient cells.
Collapse
Affiliation(s)
- Yang Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tianyu Yu
- Department of General Surgery, Pudong New Area Gongli Hospital Affiliated to Naval Military Medical University, Naval Military Medical University, Shanghai, China
| | - Yi Han
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yazhi He
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yiran Song
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Leiming Guo
- Department of R&D, Shanghai Creative Immune Therapeutics Co., Ltd, Shanghai, China
| | - Liwei An
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunying Yang
- Central Laboratory, Shanghai Putuo District People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feng Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China.,Central Laboratory, Shanghai Putuo District People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Bolanos-Garcia VM. On the Regulation of Mitosis by the Kinetochore, a Macromolecular Complex and Organising Hub of Eukaryotic Organisms. Subcell Biochem 2022; 99:235-267. [PMID: 36151378 DOI: 10.1007/978-3-031-00793-4_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The kinetochore is the multiprotein complex of eukaryotic organisms that is assembled on mitotic or meiotic centromeres to connect centromeric DNA with microtubules. Its function involves the coordinated action of more than 100 different proteins. The kinetochore acts as an organiser hub that establishes physical connections with microtubules and centromere-associated proteins and recruits central protein components of the spindle assembly checkpoint (SAC), an evolutionarily conserved surveillance mechanism of eukaryotic organisms that detects unattached kinetochores and destabilises incorrect kinetochore-microtubule attachments. The molecular communication between the kinetochore and the SAC is highly dynamic and tightly regulated to ensure that cells can progress towards anaphase until each chromosome is properly bi-oriented on the mitotic spindle. This is achieved through an interplay of highly cooperative interactions and concerted phosphorylation/dephosphorylation events that are organised in time and space.This contribution discusses our current understanding of the function, structure and regulation of the kinetochore, in particular, how its communication with the SAC results in the amplification of specific signals to exquisitely control the eukaryotic cell cycle. This contribution also addresses recent advances in machine learning approaches, cell imaging and proteomics techniques that have enhanced our understanding of the molecular mechanisms that ensure the high fidelity and timely segregation of the genetic material every time a cell divides as well as the current challenges in the study of this fascinating molecular machine.
Collapse
Affiliation(s)
- Victor M Bolanos-Garcia
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK.
| |
Collapse
|
37
|
Yan X, Liu SM, Liu C. Clinical Applications of Aneuploidies in Evolution of NSCLC Patients: Current Status and Application Prospect. Onco Targets Ther 2022; 15:1355-1368. [PMID: 36388157 PMCID: PMC9662021 DOI: 10.2147/ott.s380016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 10/22/2022] [Indexed: 11/11/2022] Open
Abstract
As one of the first characteristics of cancer cells, chromosomal aberrations during cell division have been well documented. Aneuploidy is a feature of most cancer cells accompanied by an elevated rate of mis-segregation of chromosomes, called chromosome instability (CIN). Aneuploidy causes ongoing karyotypic changes that contribute to tumor heterogeneity, drug resistance, and treatment failure, which are considered predictors of poor prognosis. Lung cancer (LC) is the leading cause of cancer-related deaths worldwide, and its genome map shows extensive aneuploid changes. Elucidating the role of aneuploidy in the pathogenesis of LC will reveal information about the key factors of tumor occurrence and development, help to predict the prognosis of cancer, clarify tumor evolution, metastasis, and drug response, and may promote the development of precision oncology. In this review, we describe many possible causes of aneuploidy and provide evidence of the role of aneuploidy in the evolution of LC, providing a basis for future biological and clinical research.
Collapse
Affiliation(s)
- Xing Yan
- The Second Affiliated Hospital of Dalian Medical University, Dalian, 116000, People's Republic of China
| | - Shan Mei Liu
- Inner Mongolia Medical University, Hohhot, 150110, People's Republic of China
| | - Changhong Liu
- The Second Affiliated Hospital of Dalian Medical University, Dalian, 116000, People's Republic of China
| |
Collapse
|
38
|
Jung Y, Kraikivski P, Shafiekhani S, Terhune SS, Dash RK. Crosstalk between Plk1, p53, cell cycle, and G2/M DNA damage checkpoint regulation in cancer: computational modeling and analysis. NPJ Syst Biol Appl 2021; 7:46. [PMID: 34887439 PMCID: PMC8660825 DOI: 10.1038/s41540-021-00203-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/03/2021] [Indexed: 12/21/2022] Open
Abstract
Different cancer cell lines can have varying responses to the same perturbations or stressful conditions. Cancer cells that have DNA damage checkpoint-related mutations are often more sensitive to gene perturbations including altered Plk1 and p53 activities than cancer cells without these mutations. The perturbations often induce a cell cycle arrest in the former cancer, whereas they only delay the cell cycle progression in the latter cancer. To study crosstalk between Plk1, p53, and G2/M DNA damage checkpoint leading to differential cell cycle regulations, we developed a computational model by extending our recently developed model of mitotic cell cycle and including these key interactions. We have used the model to analyze the cancer cell cycle progression under various gene perturbations including Plk1-depletion conditions. We also analyzed mutations and perturbations in approximately 1800 different cell lines available in the Cancer Dependency Map and grouped lines by genes that are represented in our model. Our model successfully explained phenotypes of various cancer cell lines under different gene perturbations. Several sensitivity analysis approaches were used to identify the range of key parameter values that lead to the cell cycle arrest in cancer cells. Our resulting model can be used to predict the effect of potential treatments targeting key mitotic and DNA damage checkpoint regulators on cell cycle progression of different types of cancer cells.
Collapse
Affiliation(s)
- Yongwoon Jung
- grid.30760.320000 0001 2111 8460Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - Pavel Kraikivski
- Academy of Integrated Science, Division of Systems Biology, Virginia Tech, Blacksburg, VA, 24061, USA.
| | - Sajad Shafiekhani
- grid.411705.60000 0001 0166 0922Department of Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Scott S. Terhune
- grid.30760.320000 0001 2111 8460Departments of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226 USA ,grid.30760.320000 0001 2111 8460Center of Systems and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - Ranjan K. Dash
- grid.30760.320000 0001 2111 8460Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226 USA ,grid.30760.320000 0001 2111 8460Center of Systems and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226 USA ,grid.30760.320000 0001 2111 8460Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| |
Collapse
|
39
|
Wang C, Chen D, Pan C, Wang C. Research progress of Bub3 gene in malignant tumors. Cell Biol Int 2021; 46:673-682. [PMID: 34882895 PMCID: PMC9303375 DOI: 10.1002/cbin.11740] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/28/2021] [Accepted: 12/04/2021] [Indexed: 12/09/2022]
Abstract
The spindle assembly checkpoint (SAC) is a highly conserved monitoring system that ensures a fidelity of chromosome segregation during mitosis. Bub3, a mitotic Checkpoint Protein, is a member of the Bub protein family, and an important factor in the SAC. Abnormal expression of Bub3 results in mitotic defects, defective spindle gate function, chromosomal instability and the development of aneuploidy cells. Aneuploidy is a state of abnormal karyotype that has long been considered as a marker of tumorigenesis. Karyotypic heterogeneity in tumor cells, known as "chromosomal instability" (CIN), can be used to distinguish cancerous cells from their normal tissue counterpart. In this review, we summarize the expression and clinical significance of Bub3 in a variety of tumors and suggest that it has potential in the treatment of cancer. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Chenyang Wang
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.,1997-09, Woman, Han, Breast cancer
| | - Dating Chen
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Chenglong Pan
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Chunyan Wang
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.,1974-07, Woman, Han, Breast cancer
| |
Collapse
|
40
|
Choi BH, Colon TM, Lee E, Kou Z, Dai W. CBX8 interacts with chromatin PTEN and is involved in regulating mitotic progression. Cell Prolif 2021; 54:e13110. [PMID: 34592789 PMCID: PMC8560621 DOI: 10.1111/cpr.13110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/14/2021] [Accepted: 07/27/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Besides its role in regulating phosphatidylinositol-3 kinase (PI3K) signalling in the cytosol, PTEN also has a nuclear function. In this study, we attempted to understand the mechanism of chromatin PTEN in suppressing chromosomal instability during cell division. MATERIALS AND METHODS Immunocoprecipitation, ectopic expression, and deletional analyses were used to identify the physical interaction between Chromobox Homolog protein 8 (CBX8) and PTEN, as well as the functional domain(s) of PTEN mediating the interaction. Cell synchronization followed by immunoblotting was employed to study cell cycle regulation of CBX8 and the functional interaction between chromatin PTEN and CBX8. Small interfering RNAs (siRNAs) were used to study the role of PTEN and CBX8 in modulating histone epigenetic markers during the cell cycle. RESULTS Polycomb group (PcG) proteins including CBXs function to repress gene expression in a wide range of organisms including mammals. We recently showed that PTEN interacted with CBX8, a component of Polycomb Repressing Complex 1 (PRC1), and that CBX8 co-localized with PTEN in the nucleus. CBX8 levels were high, coinciding with its phosphorylation in mitosis. Phosphorylation of CBX8 was associated with monoubiquitinated PTEN and phosphorylated-BubR1 on chromatin. Moreover, CBX8 played an important role in cell proliferation and mitotic progression. Significantly, downregulation of either PTEN or CBX8 induced H3K27Me3 epigenetic marker in mitotic cells. CONCLUSION CBX8 is a new component that physically interacts with chromatin PTEN, playing an important role in regulating mitotic progression.
Collapse
Affiliation(s)
- Byeong Hyeok Choi
- Department of Environmental MedicineNew York University Grossman School of MedicineNew YorkNYUSA
| | - Tania Marlyn Colon
- Department of Environmental MedicineNew York University Grossman School of MedicineNew YorkNYUSA
| | - Eunji Lee
- Department of Environmental MedicineNew York University Grossman School of MedicineNew YorkNYUSA
| | - Ziyue Kou
- Department of Environmental MedicineNew York University Grossman School of MedicineNew YorkNYUSA
| | - Wei Dai
- Department of Environmental MedicineNew York University Grossman School of MedicineNew YorkNYUSA
- Department of Biochemistry and Molecular PharmacologyNew York University Langone Medical CenterNew YorkNYUSA
| |
Collapse
|
41
|
Deng DJ, Xia QC, Jia GS, Suo F, Chen JL, Sun L, Wang JQ, Wang SM, Du LL, Wang Y, Jin QW. Perturbation of kinetochore function using GFP-binding protein in fission yeast. G3 GENES|GENOMES|GENETICS 2021; 11:6353032. [PMID: 34849791 PMCID: PMC8527488 DOI: 10.1093/g3journal/jkab290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/10/2021] [Indexed: 11/12/2022]
Abstract
Abstract
Using genetic mutations to study protein functions in vivo is a central paradigm of modern biology. Single-domain camelid antibodies generated against GFP have been engineered as nanobodies or GFP-binding proteins (GBPs) that can bind GFP as well as some GFP variants with high affinity and selectivity. In this study, we have used GBP-mCherry fusion protein as a tool to perturb the natural functions of a few kinetochore proteins in the fission yeast Schizosaccharomyces pombe. We found that cells simultaneously expressing GBP-mCherry and the GFP-tagged inner kinetochore protein Cnp1 are sensitive to high temperature and microtubule drug thiabendazole (TBZ). In addition, kinetochore-targeted GBP-mCherry by a few major kinetochore proteins with GFP tags causes defects in faithful chromosome segregation. Thus, this setting compromises the functions of kinetochores and renders cells to behave like conditional mutants. Our study highlights the potential of using GBP as a general tool to perturb the function of some GFP-tagged proteins in vivo with the objective of understanding their functional relevance to certain physiological processes, not only in yeasts, but also potentially in other model systems.
Collapse
Affiliation(s)
- Da-Jie Deng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Qian-Cheng Xia
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Guo-Song Jia
- National Institute of Biological Sciences, Beijing 102206, China
| | - Fang Suo
- National Institute of Biological Sciences, Beijing 102206, China
| | - Jia-Li Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Li Sun
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Jin-Qing Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Shuang-Min Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Li-Lin Du
- National Institute of Biological Sciences, Beijing 102206, China
| | - Yamei Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Quan-Wen Jin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| |
Collapse
|
42
|
Komura K, Inamoto T, Tsujino T, Matsui Y, Konuma T, Nishimura K, Uchimoto T, Tsutsumi T, Matsunaga T, Maenosono R, Yoshikawa Y, Taniguchi K, Tanaka T, Uehara H, Hirata K, Hirano H, Nomi H, Hirose Y, Ono F, Azuma H. Increased BUB1B/BUBR1 expression contributes to aberrant DNA repair activity leading to resistance to DNA-damaging agents. Oncogene 2021; 40:6210-6222. [PMID: 34545188 PMCID: PMC8553621 DOI: 10.1038/s41388-021-02021-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 08/24/2021] [Accepted: 09/10/2021] [Indexed: 11/16/2022]
Abstract
There has been accumulating evidence for the clinical benefit of chemoradiation therapy (CRT), whereas mechanisms in CRT-recurrent clones derived from the primary tumor are still elusive. Herein, we identified an aberrant BUB1B/BUBR1 expression in CRT-recurrent clones in bladder cancer (BC) by comprehensive proteomic analysis. CRT-recurrent BC cells exhibited a cell-cycle-independent upregulation of BUB1B/BUBR1 expression rendering an enhanced DNA repair activity in response to DNA double-strand breaks (DSBs). With DNA repair analyses employing the CRISPR/cas9 system, we revealed that cells with aberrant BUB1B/BUBR1 expression dominantly exploit mutagenic nonhomologous end joining (NHEJ). We further found that phosphorylated ATM interacts with BUB1B/BUBR1 after ionizing radiation (IR) treatment, and the resistance to DSBs by increased BUB1B/BUBR1 depends on the functional ATM. In vivo, tumor growth of CRT-resistant T24R cells was abrogated by ATM inhibition using AZD0156. A dataset analysis identified FOXM1 as a putative BUB1B/BUBR1-targeting transcription factor causing its increased expression. These data collectively suggest a redundant role of BUB1B/BUBR1 underlying mutagenic NHEJ in an ATM-dependent manner, aside from the canonical activity of BUB1B/BUBR1 on the G2/M checkpoint, and offer novel clues to overcome CRT resistance.
Collapse
Affiliation(s)
- Kazumasa Komura
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan. .,Translational Research Program, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan.
| | - Teruo Inamoto
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| | - Takuya Tsujino
- Division of Urology, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, United States
| | - Yusuke Matsui
- Biomedical and Health Informatics Unit, Department of Integrated Health Science, Nagoya University Graduate School of Medicine, Nagoya, 461-8673, Japan.,Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, 461-8673, Japan
| | - Tsuyoshi Konuma
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan
| | - Kazuki Nishimura
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| | - Taizo Uchimoto
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| | - Takeshi Tsutsumi
- Division of Urology, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, United States
| | - Tomohisa Matsunaga
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| | - Ryoichi Maenosono
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| | - Yuki Yoshikawa
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| | - Kohei Taniguchi
- Translational Research Program, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| | - Tomohito Tanaka
- Translational Research Program, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| | - Hirofumi Uehara
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| | - Koichi Hirata
- Department of Pathology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| | - Hajime Hirano
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| | - Hayahito Nomi
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| | - Yoshinobu Hirose
- Department of Pathology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| | - Fumihito Ono
- Translational Research Program, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan.,Department of Physiology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| | - Haruhito Azuma
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan
| |
Collapse
|
43
|
Lara-Gonzalez P, Pines J, Desai A. Spindle assembly checkpoint activation and silencing at kinetochores. Semin Cell Dev Biol 2021; 117:86-98. [PMID: 34210579 PMCID: PMC8406419 DOI: 10.1016/j.semcdb.2021.06.009] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/17/2021] [Accepted: 06/17/2021] [Indexed: 01/01/2023]
Abstract
The spindle assembly checkpoint (SAC) is a surveillance mechanism that promotes accurate chromosome segregation in mitosis. The checkpoint senses the attachment state of kinetochores, the proteinaceous structures that assemble onto chromosomes in mitosis in order to mediate their interaction with spindle microtubules. When unattached, kinetochores generate a diffusible inhibitor that blocks the activity of the anaphase-promoting complex/cyclosome (APC/C), an E3 ubiquitin ligase required for sister chromatid separation and exit from mitosis. Work from the past decade has greatly illuminated our understanding of the mechanisms by which the diffusible inhibitor is assembled and how it inhibits the APC/C. However, less is understood about how SAC proteins are recruited to kinetochores in the absence of microtubule attachment, how the kinetochore catalyzes formation of the diffusible inhibitor, and how attachments silence the SAC at the kinetochore. Here, we summarize current understanding of the mechanisms that activate and silence the SAC at kinetochores and highlight open questions for future investigation.
Collapse
Affiliation(s)
- Pablo Lara-Gonzalez
- Ludwig Institute for Cancer Research, USA; Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| | | | - Arshad Desai
- Ludwig Institute for Cancer Research, USA; Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
44
|
Kops GJPL, Snel B, Tromer EC. Evolutionary Dynamics of the Spindle Assembly Checkpoint in Eukaryotes. Curr Biol 2021; 30:R589-R602. [PMID: 32428500 DOI: 10.1016/j.cub.2020.02.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The tremendous diversity in eukaryotic life forms can ultimately be traced back to evolutionary modifications at the level of molecular networks. Deep understanding of these modifications will not only explain cellular diversity, but will also uncover different ways to execute similar processes and expose the evolutionary 'rules' that shape the molecular networks. Here, we review the evolutionary dynamics of the spindle assembly checkpoint (SAC), a signaling network that guards fidelity of chromosome segregation. We illustrate how the interpretation of divergent SAC systems in eukaryotic species is facilitated by combining detailed molecular knowledge of the SAC and extensive comparative genome analyses. Ultimately, expanding this to other core cellular systems and experimentally interrogating such systems in organisms from all major lineages may start outlining the routes to and eventual manifestation of the cellular diversity of eukaryotic life.
Collapse
Affiliation(s)
- Geert J P L Kops
- Oncode Institute, Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Centre Utrecht, Utrecht, The Netherlands.
| | - Berend Snel
- Theoretical Biology and Bioinformatics, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands.
| | - Eelco C Tromer
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
| |
Collapse
|
45
|
Thomas C, Wetherall B, Levasseur MD, Harris RJ, Kerridge ST, Higgins JMG, Davies OR, Madgwick S. A prometaphase mechanism of securin destruction is essential for meiotic progression in mouse oocytes. Nat Commun 2021; 12:4322. [PMID: 34262048 PMCID: PMC8280194 DOI: 10.1038/s41467-021-24554-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 06/24/2021] [Indexed: 11/10/2022] Open
Abstract
Successful cell division relies on the timely removal of key cell cycle proteins such as securin. Securin inhibits separase, which cleaves the cohesin rings holding chromosomes together. Securin must be depleted before anaphase to ensure chromosome segregation occurs with anaphase. Here we find that in meiosis I, mouse oocytes contain an excess of securin over separase. We reveal a mechanism that promotes excess securin destruction in prometaphase I. Importantly, this mechanism relies on two phenylalanine residues within the separase-interacting segment (SIS) of securin that are only exposed when securin is not bound to separase. We suggest that these residues facilitate the removal of non-separase-bound securin ahead of metaphase, as inhibiting this period of destruction by mutating both residues causes the majority of oocytes to arrest in meiosis I. We further propose that cellular securin levels exceed the amount an oocyte is capable of removing in metaphase alone, such that the prometaphase destruction mechanism identified here is essential for correct meiotic progression in mouse oocytes.
Collapse
Affiliation(s)
- Christopher Thomas
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK. .,Max Planck Institute for Biophysical Chemistry, Gottingen, Germany.
| | - Benjamin Wetherall
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mark D Levasseur
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Rebecca J Harris
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Scott T Kerridge
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Jonathan M G Higgins
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Owen R Davies
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh, UK
| | - Suzanne Madgwick
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
46
|
Lambhate S, Bhattacharjee D, Jain N. APC/C CDH1 ubiquitinates IDH2 contributing to ROS increase in mitosis. Cell Signal 2021; 86:110087. [PMID: 34271087 DOI: 10.1016/j.cellsig.2021.110087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 11/15/2022]
Abstract
NADPH is a cofactor used by reactive oxygen species (ROS) scavenging enzymes to block ROS produced in cells. Recently, it was shown that in cancer cells, ROS progressively increases in tune to cell cycle leading to a peak in mitosis. Loss of IDH2 is known to cause severe oxidative stress in cell and mouse models as ROS increases in mitochondria. Therefore, we hypothesized that IDH2, a major NADPH-producing enzyme in mitochondria is ubiquitinated for ROS to increase in mitosis. To test this hypothesis, in cancer cells we examined IDH2 ubiquitination in mitosis and measured the ROS produced. We found that IDH2 is ubiquitinated in mitosis and on inhibiting anaphase-promoting complex/Cyclosome (APC/C) IDH2 was stabilized. Further, we observed that overexpressing APC/C coactivator CDH1 decreased IDH2, whereas depleting CDH1 decreased IDH2 ubiquitination. To understand the link between IDH2 ubiquitination and ROS produced in mitosis, we show that overexpressing mitochondria-targeted-IDH1 decreased ROS by increasing NADPH in IDH2 ubiquitinated cells. We conclude that APC/C CDH1 ubiquitinates IDH2, a major NADPH-producing enzyme in mitochondria contributing to ROS increase in mitosis. Based on our results, we suggest that mitosis can be a therapeutic window in mutant IDH2-linked pathologies.
Collapse
Affiliation(s)
- Surbhi Lambhate
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Debanjan Bhattacharjee
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Nishant Jain
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
47
|
Fischer ES, Yu CWH, Bellini D, McLaughlin SH, Orr CM, Wagner A, Freund SMV, Barford D. Molecular mechanism of Mad1 kinetochore targeting by phosphorylated Bub1. EMBO Rep 2021; 22:e52242. [PMID: 34013668 PMCID: PMC8391104 DOI: 10.15252/embr.202052242] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/02/2021] [Accepted: 04/13/2021] [Indexed: 01/10/2023] Open
Abstract
During metaphase, in response to improper kinetochore-microtubule attachments, the spindle assembly checkpoint (SAC) activates the mitotic checkpoint complex (MCC), an inhibitor of the anaphase-promoting complex/cyclosome (APC/C). This process is orchestrated by the kinase Mps1, which initiates the assembly of the MCC onto kinetochores through a sequential phosphorylation-dependent signalling cascade. The Mad1-Mad2 complex, which is required to catalyse MCC formation, is targeted to kinetochores through a direct interaction with the phosphorylated conserved domain 1 (CD1) of Bub1. Here, we present the crystal structure of the C-terminal domain of Mad1 (Mad1CTD ) bound to two phosphorylated Bub1CD1 peptides at 1.75 Å resolution. This interaction is mediated by phosphorylated Bub1 Thr461, which not only directly interacts with Arg617 of the Mad1 RLK (Arg-Leu-Lys) motif, but also directly acts as an N-terminal cap to the CD1 α-helix dipole. Surprisingly, only one Bub1CD1 peptide binds to the Mad1 homodimer in solution. We suggest that this stoichiometry is due to inherent asymmetry in the coiled-coil of Mad1CTD and has implications for how the Mad1-Bub1 complex at kinetochores promotes efficient MCC assembly.
Collapse
Affiliation(s)
| | | | - Dom Bellini
- MRC Laboratory of Molecular BiologyCambridgeUK
| | | | | | | | | | | |
Collapse
|
48
|
Koliopoulos MG, Alfieri C. Cell cycle regulation by complex nanomachines. FEBS J 2021; 289:5100-5120. [PMID: 34143558 DOI: 10.1111/febs.16082] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/05/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022]
Abstract
The cell cycle is the essential biological process where one cell replicates its genome and segregates the resulting two copies into the daughter cells during mitosis. Several aspects of this process have fascinated humans since the nineteenth century. Today, the cell cycle is exhaustively investigated because of its profound connections with human diseases and cancer. At the heart of the molecular network controlling the cell cycle, we find the cyclin-dependent kinases (CDKs) acting as an oscillator to impose an orderly and highly regulated progression through the different cell cycle phases. This oscillator integrates both internal and external signals via a multitude of signalling pathways involving posttranslational modifications including phosphorylation, protein ubiquitination and mechanisms of transcriptional regulation. These tasks are specifically performed by multi-subunit complexes, which are intensively studied both biochemically and structurally with the aim to unveil mechanistic insights into their molecular function. The scope of this review is to summarise the structural biology of the cell cycle machinery, with specific focus on the core cell cycle machinery involving the CDK-cyclin oscillator. We highlight the contribution of cryo-electron microscopy, which has started to revolutionise our understanding of the molecular function and dynamics of the key players of the cell cycle.
Collapse
Affiliation(s)
- Marios G Koliopoulos
- Chester Beatty Laboratories, Structural Biology Division, Institute of Cancer Research, London, UK
| | - Claudio Alfieri
- Chester Beatty Laboratories, Structural Biology Division, Institute of Cancer Research, London, UK
| |
Collapse
|
49
|
Bodrug T, Welsh KA, Hinkle M, Emanuele MJ, Brown NG. Intricate Regulatory Mechanisms of the Anaphase-Promoting Complex/Cyclosome and Its Role in Chromatin Regulation. Front Cell Dev Biol 2021; 9:687515. [PMID: 34109183 PMCID: PMC8182066 DOI: 10.3389/fcell.2021.687515] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/26/2021] [Indexed: 02/04/2023] Open
Abstract
The ubiquitin (Ub)-proteasome system is vital to nearly every biological process in eukaryotes. Specifically, the conjugation of Ub to target proteins by Ub ligases, such as the Anaphase-Promoting Complex/Cyclosome (APC/C), is paramount for cell cycle transitions as it leads to the irreversible destruction of cell cycle regulators by the proteasome. Through this activity, the RING Ub ligase APC/C governs mitosis, G1, and numerous aspects of neurobiology. Pioneering cryo-EM, biochemical reconstitution, and cell-based studies have illuminated many aspects of the conformational dynamics of this large, multi-subunit complex and the sophisticated regulation of APC/C function. More recent studies have revealed new mechanisms that selectively dictate APC/C activity and explore additional pathways that are controlled by APC/C-mediated ubiquitination, including an intimate relationship with chromatin regulation. These tasks go beyond the traditional cell cycle role historically ascribed to the APC/C. Here, we review these novel findings, examine the mechanistic implications of APC/C regulation, and discuss the role of the APC/C in previously unappreciated signaling pathways.
Collapse
Affiliation(s)
- Tatyana Bodrug
- Department of Biochemistry and Biophysics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kaeli A Welsh
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Megan Hinkle
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Michael J Emanuele
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Nicholas G Brown
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| |
Collapse
|
50
|
Hein JB, Garvanska DH, Nasa I, Kettenbach AN, Nilsson J. Coupling of Cdc20 inhibition and activation by BubR1. J Cell Biol 2021; 220:211939. [PMID: 33819340 PMCID: PMC8025235 DOI: 10.1083/jcb.202012081] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/18/2021] [Accepted: 02/25/2021] [Indexed: 12/25/2022] Open
Abstract
Tight regulation of the APC/C-Cdc20 ubiquitin ligase that targets cyclin B1 for degradation is important for mitotic fidelity. The spindle assembly checkpoint (SAC) inhibits Cdc20 through the mitotic checkpoint complex (MCC). In addition, phosphorylation of Cdc20 by cyclin B1–Cdk1 independently inhibits APC/C–Cdc20 activation. This creates a conundrum for how Cdc20 is activated before cyclin B1 degradation. Here, we show that the MCC component BubR1 harbors both Cdc20 inhibition and activation activities, allowing for cross-talk between the two Cdc20 inhibition pathways. Specifically, BubR1 acts as a substrate specifier for PP2A-B56 to enable efficient Cdc20 dephosphorylation in the MCC. A mutant Cdc20 mimicking the dephosphorylated state escapes a mitotic checkpoint arrest, arguing that restricting Cdc20 dephosphorylation to the MCC is important. Collectively, our work reveals how Cdc20 can be dephosphorylated in the presence of cyclin B1-Cdk1 activity without causing premature anaphase onset.
Collapse
Affiliation(s)
- Jamin B Hein
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Science, Copenhagen, Denmark
| | - Dimitriya H Garvanska
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Science, Copenhagen, Denmark
| | - Isha Nasa
- Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH
| | - Arminja N Kettenbach
- Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH
| | - Jakob Nilsson
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Science, Copenhagen, Denmark
| |
Collapse
|